Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Nat Immunol ; 21(6): 696, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32210390

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Nat Immunol ; 20(6): 756-764, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31110315

RESUMO

Emerging data show that tissue-resident memory T (TRM) cells play an important protective role at murine and human barrier sites. TRM cells in the epidermis of mouse skin patrol their surroundings and rapidly respond when antigens are encountered. However, whether a similar migratory behavior is performed by human TRM cells is unclear, as technology to longitudinally follow them in situ has been lacking. To address this issue, we developed an ex vivo culture system to label and track T cells in fresh skin samples. We validated this system by comparing in vivo and ex vivo properties of murine TRM cells. Using nanobody labeling, we subsequently demonstrated in human ex vivo skin that CD8+ TRM cells migrated through the papillary dermis and the epidermis, below sessile Langerhans cells. Collectively, this work allows the dynamic study of resident immune cells in human skin and provides evidence of tissue patrol by human CD8+ TRM cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Pele/imunologia , Animais , Antígenos/imunologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Epiderme/imunologia , Epiderme/metabolismo , Imunofluorescência , Humanos , Camundongos , Especificidade de Órgãos/imunologia , Anticorpos de Domínio Único/imunologia , Pele/metabolismo , Vacinas de DNA/genética , Vacinas de DNA/imunologia
3.
Mol Cell Proteomics ; 19(7): 1179-1192, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32332107

RESUMO

The vessel wall is continuously exposed to hemodynamic forces generated by blood flow. Endothelial mechanosensors perceive and translate mechanical signals via cellular signaling pathways into biological processes that control endothelial development, phenotype and function. To assess the hemodynamic effects on the endothelium on a system-wide level, we applied a quantitative mass spectrometry approach combined with cell surface chemical footprinting. SILAC-labeled endothelial cells were subjected to flow-induced shear stress for 0, 24 or 48 h, followed by chemical labeling of surface proteins using a non-membrane permeable biotin label, and analysis of the whole proteome and the cell surface proteome by LC-MS/MS analysis. These studies revealed that of the >5000 quantified proteins 104 were altered, which were highly enriched for extracellular matrix proteins and proteins involved in cell-matrix adhesion. Cell surface proteomics indicated that LAMA4 was proteolytically processed upon flow-exposure, which corresponded to the decreased LAMA4 mass observed on immunoblot. Immunofluorescence microscopy studies highlighted that the endothelial basement membrane was drastically remodeled upon flow exposure. We observed a network-like pattern of LAMA4 and LAMA5, which corresponded to the localization of laminin-adhesion molecules ITGA6 and ITGB4. Furthermore, the adaptation to flow-exposure did not affect the inflammatory response to tumor necrosis factor α, indicating that inflammation and flow trigger fundamentally distinct endothelial signaling pathways with limited reciprocity and synergy. Taken together, this study uncovers the blood flow-induced remodeling of the basement membrane and stresses the importance of the subendothelial basement membrane in vascular homeostasis.


Assuntos
Membrana Basal/metabolismo , Circulação Sanguínea , Células Endoteliais/metabolismo , Integrinas/metabolismo , Laminina/metabolismo , Circulação Sanguínea/fisiologia , Células Cultivadas , Cromatografia Líquida , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Matriz Extracelular/metabolismo , Imunofluorescência , Perfilação da Expressão Gênica , Ontologia Genética , Hemodinâmica , Humanos , Cadeias alfa de Integrinas/metabolismo , Integrina alfa6/metabolismo , Cadeias beta de Integrinas/metabolismo , Integrina beta4/metabolismo , Mapas de Interação de Proteínas/fisiologia , Proteômica , Espectrometria de Massas em Tandem , Fator de Necrose Tumoral alfa/farmacologia
4.
Eur J Immunol ; 49(4): 576-589, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30707456

RESUMO

The BM serves as a blood-forming organ, but also supports the maintenance and immune surveillance function of many T cells. Yet, in contrast to other organs, little is known about the molecular mechanisms that drive T-cell migration to and localization inside the BM. As BM accumulates many CXCR3-expressing memory CD8+ T cells, we tested the involvement of this chemokine receptor, but found that CXCR3 is not required for BM entry. In contrast, we could demonstrate that CXCR4, which is highly expressed on both naive and memory CD8+ T cells in BM, is critically important for homing of all CD8+ T-cell subsets to the BM in mice. Upon entry into the BM parenchyma, both naïve and memory CD8+ T cells locate close to sinusoidal vessels. Intravital imaging experiments revealed that CD8 T cells are surprisingly immobile and we found that they interact with ICAM-1+VCAM-1+BP-1+ perivascular stromal cells. These cells are the major source of CXCL12, but also express key survival factors and maintenance cytokines IL-7 and IL-15. We therefore conclude that CXCR4 is not only crucial for entry of CD8+ T cells into the BM, but also controls their subsequent localization toward BM niches that support their survival.


Assuntos
Medula Óssea/imunologia , Medula Óssea/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/imunologia , Microambiente Celular , Receptores CXCR4/metabolismo , Animais , Medula Óssea/irrigação sanguínea , Medula Óssea/patologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Comunicação Celular/imunologia , Microambiente Celular/genética , Microambiente Celular/imunologia , Citocinas/biossíntese , Memória Imunológica , Camundongos , Receptores CXCR3 , Células Estromais/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
5.
Haematologica ; 105(12): 2746-2756, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33256374

RESUMO

Bone marrow endothelium plays an important role in the homing of hematopoietic stem and progenitor cells upon transplantation, but surprisingly little is known on how the bone marrow endothelial cells regulate local permeability and hematopoietic stem and progenitor cells transmigration. We show that temporal loss of vascular endothelial-cadherin function promotes vascular permeability in BM, even upon low-dose irradiation. Loss of vascular endothelial-cadherin function also enhances homing of transplanted hematopoietic stem and progenitor cells to the bone marrow of irradiated mice although engraftment is not increased. Intriguingly, stabilizing junctional vascular endothelial-cadherin in vivo reduced bone marrow permeability, but did not prevent hematopoietic stem and progenitor cells migration into the bone marrow, suggesting that hematopoietic stem and progenitor cells use the transcellular migration route to enter the bone marrow. Indeed, using an in vitro migration assay, we show that human hematopoietic stem and progenitor cells predominantly cross bone marrow endothelium in a transcellular manner in homeostasis by inducing podosome-like structures. Taken together, vascular endothelial-cadherin is crucial for BM vascular homeostasis but dispensable for the homing of hematopoietic stem and progenitor cells. These findings are important in the development of potential therapeutic targets to improve hematopoietic stem and progenitor cell homing strategies.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Podossomos , Animais , Medula Óssea , Células da Medula Óssea , Movimento Celular , Células Endoteliais , Endotélio , Células-Tronco Hematopoéticas , Camundongos , Camundongos Endogâmicos C57BL
6.
Haematologica ; 105(6): 1677-1685, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31467128

RESUMO

Bleeding disorders and thrombotic complications are major causes of morbidity and mortality with many cases being unexplained. Thrombus formation involves aberrant expression and activation of tissue factor (TF) in vascular endothelial and smooth muscle cells. Here, we sought to identify factors that modulate TF gene expression and activity in these vascular cells. The LIM-only protein FHL2 is a scaffolding protein that modulates signal transduction pathways with crucial functions in endothelial and smooth muscle cells. However, the role of FHL2 in TF regulation and thrombosis remains unexplored. Using a murine model of venous thrombosis in mesenteric vessels, we demonstrated that FHL2 deficiency results in exacerbated thrombus formation. Gain- and loss-of-function experiments revealed that FHL2 represses TF expression in endothelial and smooth muscle cells through inhibition of the transcription factors nuclear factor κB and activating protein-1. Furthermore, we observed that FHL2 interacts with the cytoplasmic tail of TF. In line with our in vivo observations, FHL2 decreases TF activity in endothelial and smooth muscle cells whereas FHL2 knockdown or deficiency results in enhanced TF activity. Finally, the FHL2 single nucleotide polymorphism rs4851770 was associated with the risk of venous thrombosis in a large population of venous thrombosis cases and control subjects from 12 studies (INVENT consortium). Altogether, our results highlight functional involvement of FHL2 in TF-mediated coagulation and identify FHL2 as a novel gene associated with venous thrombosis in humans.


Assuntos
Tromboplastina , Trombose Venosa , Animais , Variação Genética , Humanos , Proteínas com Homeodomínio LIM/genética , Camundongos , Proteínas Musculares/genética , Tromboplastina/genética , Fatores de Transcrição/genética , Trombose Venosa/genética
7.
J Immunol ; 200(5): 1790-1801, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29386254

RESUMO

Leukocyte transendothelial migration is key to inflammation. Leukocytes first start rolling over the inflamed endothelium, followed by firmly adhering to it. Under inflammatory conditions, endothelial cells express small finger-like protrusions that stick out into the lumen. The function and regulation of these structures are unclear. We present evidence that these ICAM-1- and F-actin-rich endothelial finger-like protrusions are filopodia and function as adhesive structures for leukocytes to transit from rolling to crawling but are dispensable for diapedesis. Mechanistically, these structures require the motor function of myosin-X, activity of the small GTPase Cdc42, and p21-activated kinase 4. Moreover, myosin-X expression is under control of TNF-α-mediated c-Jun N-terminal kinase activity and is upregulated in human atherosclerotic regions. To our knowledge, this is the first study to identify that regulation of endothelial filopodia is crucial for leukocyte extravasation, in particular for the initiation of leukocyte adhesion under flow conditions.


Assuntos
Células Endoteliais/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/metabolismo , Miosinas/metabolismo , Pseudópodes/metabolismo , Actinas/metabolismo , Adesão Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Endotélio Vascular/metabolismo , Células HL-60 , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Transdução de Sinais/fisiologia , Migração Transendotelial e Transepitelial/fisiologia , Regulação para Cima/fisiologia
8.
J Immunol ; 196(1): 72-9, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26597010

RESUMO

Multiple sclerosis (MS) is a chronic demyelinating disorder of the CNS characterized by immune cell infiltration across the brain vasculature into the brain, a process not yet fully understood. We previously demonstrated that the sphingolipid metabolism is altered in MS lesions. In particular, acid sphingomyelinase (ASM), a critical enzyme in the production of the bioactive lipid ceramide, is involved in the pathogenesis of MS; however, its role in the brain vasculature remains unknown. Transmigration of T lymphocytes is highly dependent on adhesion molecules in the vasculature such as intercellular adhesion molecule-1 (ICAM-1). In this article, we hypothesize that ASM controls T cell migration by regulating ICAM-1 function. To study the role of endothelial ASM in transmigration, we generated brain endothelial cells lacking ASM activity using a lentiviral shRNA approach. Interestingly, although ICAM-1 expression was increased in cells lacking ASM activity, we measured a significant decrease in T lymphocyte adhesion and consequently transmigration both in static and under flow conditions. As an underlying mechanism, we revealed that upon lack of endothelial ASM activity, the phosphorylation of ezrin was perturbed as well as the interaction between filamin and ICAM-1 upon ICAM-1 clustering. Functionally this resulted in reduced microvilli formation and impaired transendothelial migration of T cells. In conclusion, in this article, we show that ASM coordinates ICAM-1 function in brain endothelial cells by regulating its interaction with filamin and phosphorylation of ezrin. The understanding of these underlying mechanisms of T lymphocyte transmigration is of great value to develop new strategies against MS lesion formation.


Assuntos
Encéfalo/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Linfócitos T/imunologia , Migração Transendotelial e Transepitelial/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Encéfalo/citologia , Encéfalo/imunologia , Adesão Celular/genética , Adesão Celular/imunologia , Linhagem Celular , Ceramidas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Feminino , Filaminas/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/imunologia , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Fosforilação/genética , Esfingomielina Fosfodiesterase/genética , Esfingomielina Fosfodiesterase/imunologia
9.
J Immunol ; 196(3): 1272-83, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26718340

RESUMO

Neutrophils are known to play a pivotal role in the host defense against Aspergillus infections. This is illustrated by the prevalence of Aspergillus infections in patients with neutropenia or phagocyte functional defects, such as chronic granulomatous disease. However, the mechanisms by which human neutrophils recognize and kill Aspergillus are poorly understood. In this work, we have studied in detail which neutrophil functions, including neutrophil extracellular trap (NET) formation, are involved in the killing of Aspergillus fumigatus conidia and hyphae, using neutrophils from patients with well-defined genetic immunodeficiencies. Recognition of conidia involves integrin CD11b/CD18 (and not dectin-1), which triggers a PI3K-dependent nonoxidative intracellular mechanism of killing. When the conidia escape from early killing and germinate, the extracellular destruction of the Aspergillus hyphae needs opsonization by Abs and involves predominantly recognition via Fcγ receptors, signaling via Syk, PI3K, and protein kinase C to trigger the production of toxic reactive oxygen metabolites by the NADPH oxidase and myeloperoxidase. A. fumigatus induces NET formation; however, NETs did not contribute to A. fumigatus killing. Thus, our findings reveal distinct killing mechanisms of Aspergillus conidia and hyphae by human neutrophils, leading to a comprehensive insight in the innate antifungal response.


Assuntos
Aspergilose/imunologia , Aspergillus fumigatus/imunologia , Hifas/imunologia , Neutrófilos/imunologia , Esporos Fúngicos/imunologia , Citotoxicidade Imunológica/imunologia , Armadilhas Extracelulares/imunologia , Humanos , Imunidade Inata , Síndromes de Imunodeficiência/imunologia , Microscopia Confocal , Fagócitos/imunologia
10.
J Immunol ; 197(11): 4312-4324, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27815426

RESUMO

Dendritic cells (DCs) play a pivotal role in the regulation of the immune response. DC development and activation is finely orchestrated through transcriptional programs. GATA1 transcription factor is required for murine DC development, and data suggest that it might be involved in the fine-tuning of the life span and function of activated DCs. We generated DC-specific Gata1 knockout mice (Gata1-KODC), which presented a 20% reduction of splenic DCs, partially explained by enhanced apoptosis. RNA sequencing analysis revealed a number of deregulated genes involved in cell survival, migration, and function. DC migration toward peripheral lymph nodes was impaired in Gata1-KODC mice. Migration assays performed in vitro showed that this defect was selective for CCL21, but not CCL19. Interestingly, we show that Gata1-KODC DCs have reduced polysialic acid levels on their surface, which is a known determinant for the proper migration of DCs toward CCL21.


Assuntos
Movimento Celular/imunologia , Quimiocina CCL21/imunologia , Células Dendríticas/imunologia , Fator de Transcrição GATA1/imunologia , Linfonodos/imunologia , Ácidos Siálicos/imunologia , Animais , Movimento Celular/genética , Quimiocina CCL19/genética , Quimiocina CCL19/imunologia , Quimiocina CCL21/genética , Células Dendríticas/citologia , Fator de Transcrição GATA1/deficiência , Linfonodos/citologia , Camundongos , Camundongos Knockout , Ácidos Siálicos/genética
11.
J Cell Sci ; 128(16): 3041-54, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26116572

RESUMO

Endothelial cell-cell junctions maintain a restrictive barrier that is tightly regulated to allow dynamic responses to permeability-inducing angiogenic factors, as well as to inflammatory agents and adherent leukocytes. The ability of these stimuli to transiently remodel adherens junctions depends on Rho-GTPase-controlled cytoskeletal rearrangements. How the activity of Rho-GTPases is spatio-temporally controlled at endothelial adherens junctions by guanine-nucleotide exchange factors (GEFs) is incompletely understood. Here, we identify a crucial role for the Rho-GEF Trio in stabilizing junctions based around vascular endothelial (VE)-cadherin (also known as CDH5). Trio interacts with VE-cadherin and locally activates Rac1 at adherens junctions during the formation of nascent contacts, as assessed using a novel FRET-based Rac1 biosensor and biochemical assays. The Rac-GEF domain of Trio is responsible for the remodeling of junctional actin from radial into cortical actin bundles, a crucial step for junction stabilization. This promotes the formation of linear adherens junctions and increases endothelial monolayer resistance. Collectively, our data show the importance of spatio-temporal regulation of the actin cytoskeleton through Trio and Rac1 at VE-cadherin-based cell-cell junctions in the maintenance of the endothelial barrier.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Junções Intercelulares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Antígenos CD/genética , Caderinas/genética , Permeabilidade Capilar/genética , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Proteínas rac1 de Ligação ao GTP/genética
12.
Blood ; 125(12): 1957-67, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25538045

RESUMO

Sp1 and Sp3 belong to the specificity proteins (Sp)/Krüppel-like transcription factor family. They are closely related, ubiquitously expressed, and recognize G-rich DNA motifs. They are thought to regulate generic processes such as cell-cycle and growth control, metabolic pathways, and apoptosis. Ablation of Sp1 or Sp3 in mice is lethal, and combined haploinsufficiency results in hematopoietic defects during the fetal stages. Here, we show that in adult mice, conditional pan-hematopoietic (Mx1-Cre) ablation of either Sp1 or Sp3 has minimal impact on hematopoiesis, whereas the simultaneous loss of Sp1 and Sp3 results in severe macrothrombocytopenia. This occurs in a cell-autonomous manner as shown by megakaryocyte-specific (Pf4-Cre) double-knockout mice. We employed flow cytometry, cell culture, and electron microscopy and show that although megakaryocyte numbers are normal in bone marrow and spleen, they display a less compact demarcation membrane system and a striking inability to form proplatelets. Through megakaryocyte transcriptomics and platelet proteomics, we identified several cytoskeleton-related proteins and downstream effector kinases, including Mylk, that were downregulated upon Sp1/Sp3 depletion, providing an explanation for the observed defects in megakaryopoiesis. Supporting this notion, selective Mylk inhibition by ML7 affected proplatelet formation and stabilization and resulted in defective ITAM receptor-mediated platelet aggregation.


Assuntos
Plaquetas/citologia , Megacariócitos/citologia , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3/genética , Animais , Azepinas/química , Plaquetas/metabolismo , Medula Óssea/metabolismo , Citometria de Fluxo , Lectinas Tipo C/metabolismo , Camundongos , Camundongos Knockout , Naftalenos/química , Agregação Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Proteoma , Transdução de Sinais , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3/metabolismo , Baço/metabolismo , Trombocitopenia/metabolismo , Fatores de Transcrição/metabolismo
13.
J Cell Sci ; 127(Pt 20): 4470-82, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25107367

RESUMO

Chronic vascular inflammation is driven by interactions between activated leukocytes and the endothelium. Leukocyte ß2-integrins bind to endothelial intercellular adhesion molecule 1 (ICAM-1), which allows leukocyte spreading, crawling and transendothelial migration. Leukocytes scan the vascular endothelium for permissive sites to transmigrate, which suggests that there is apical membrane heterogeneity within the endothelium. However, the molecular basis for this heterogeneity is unknown. Leukocyte adhesion induces ICAM-1 clustering, which promotes its association to the actin-binding proteins filamin B, α-actinin-4 and cortactin. We show that these endothelial proteins differentially control adhesion, spreading and transmigration of neutrophils. Loss of filamin B, α-actinin-4 and cortactin revealed adaptor-specific effects on a nuclear-to-peripheral gradient of endothelial cell stiffness. By contrast, increasing endothelial cell stiffness stimulates ICAM-1 function. We identify endothelial α-actinin-4 as a key regulator of endothelial cell stiffness and of ICAM-1-mediated neutrophil transmigration. Finally, we found that the endothelial lining of human and murine atherosclerotic plaques shows elevated levels of α-actinin-4. These results identify endothelial cell stiffness as an important regulator of endothelial surface heterogeneity and of ICAM-1 function, which in turn controls the adhesion and transmigration of neutrophils.


Assuntos
Actinina/metabolismo , Células Endoteliais/metabolismo , Filaminas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Toxinas Marinhas/metabolismo , Neutrófilos/fisiologia , Placa Aterosclerótica/metabolismo , Migração Transendotelial e Transepitelial , Actinina/genética , Actinas/metabolismo , Animais , Adesão Celular/genética , Células Endoteliais/citologia , Filaminas/genética , Células HeLa , Humanos , Masculino , Toxinas Marinhas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/genética
14.
J Immunol ; 192(9): 4370-8, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24670805

RESUMO

Azathioprine and its metabolite 6-mercaptopurine (6-MP) are well established immunosuppressive drugs. Common understanding of their immunosuppressive properties is largely limited to immune cells. However, in this study, the mechanism underlying the protective role of 6-MP in endothelial cell activation is investigated. Because 6-MP and its derivative 6-thioguanosine-5'-triphosphate (6-T-GTP) were shown to block activation of GTPase Rac1 in T lymphocytes, we focused on Rac1-mediated processes in endothelial cells. Indeed, 6-MP and 6-T-GTP decreased Rac1 activation in endothelial cells. As a result, the compounds inhibited TNF-α-induced downstream signaling via JNK and reduced activation of transcription factors c-Jun, activating transcription factor-2 and, in addition, NF κ-light-chain-enhancer of activated B cells (NF-κB), which led to decreased transcription of proinflammatory cytokines. Moreover, 6-MP and 6-T-GTP selectively decreased TNF-α-induced VCAM-1 but not ICAM-1 protein levels. Rac1-mediated generation of cell membrane protrusions, which form docking structures to capture leukocytes, also was reduced by 6-MP/6-T-GTP. Consequently, leukocyte transmigration was inhibited after 6-MP/6-T-GTP treatment. These data underscore the anti-inflammatory effect of 6-MP and 6-T-GTP on endothelial cells by blocking Rac1 activation. Our data provide mechanistic insight that supports development of novel Rac1-specific therapeutic approaches against chronic inflammatory diseases.


Assuntos
Células Endoteliais/efeitos dos fármacos , Imunossupressores/farmacologia , Mercaptopurina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Western Blotting , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Transcriptoma , Migração Transendotelial e Transepitelial/efeitos dos fármacos
15.
Blood Cells Mol Dis ; 55(4): 293-303, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26460250

RESUMO

Dendritic cells (DCs) are key initiators and regulators of the immune response. The development of the DC lineage and their subsets requires an orchestrated regulation of their transcriptional program. Gata1, a transcription factor expressed in several hematopoietic cell lineages, has been recently reported to be required for mouse DC development and function. In humans, GATA1 is involved in the lineage separation between monocyte-derived DCs and Langerhans cells (LC) and loss of GATA1 results in differentiation arrest at the monocyte stage. The hematopoietic GATA factors (i.e. Gata1, Gata2, Gata3) are known to regulate each other's expression and to function consecutively throughout lineage commitment (so-called GATA switch). In humans, mutations in GATA2 are causative of MonoMAC disease, a human immunodeficiency syndrome characterized by loss of DCs, monocytes, B and NK cells. However, additional data on the expression of hematopoietic GATA factors in the DC lineage is missing. In this study, we have characterized the expression of hematopoietic GATA factors in murine and human DCs and their expression dynamics upon TLR stimulation. We found that all hematopoietic GATA factors are expressed in DCs, but identified species-specific differences in the relative expression of each GATA factor, and how their expression fluctuates upon stimulation.


Assuntos
Células Dendríticas/metabolismo , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica , Hematopoese/genética , Processamento Alternativo , Animais , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fatores de Transcrição GATA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-4/metabolismo , Interleucina-4/farmacologia , Camundongos , Monócitos/metabolismo , Isoformas de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 33(10): 2380-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23950142

RESUMO

OBJECTIVE: In aortic aneurysms the arterial vessel wall is dilated because of destruction of its integrity, which may lead to lethal vessel rupture. Chronic infiltration of inflammatory cells into the vessel wall is fundamental to aneurysm pathology. We aim to limit aneurysm growth by inhibition of inflammation and reducing endothelial cell (EC) activation with immunosuppressive drug azathioprine (Aza). APPROACH AND RESULTS: Aza and its metabolite 6-mercaptopurine have anti-inflammatory effects on leukocytes. We here demonstrate that treatment of ECs with 6-mercaptopurine inhibits cell activation as illustrated by reduced expression of interleukin-12, CCL5, CCL2, and vascular cell adhesion molecule-1 and inhibition of monocyte-EC adhesion. The underlying mechanism of 6-mercaptopurine involves suppression of GTPase Rac1 activation, resulting in reduced phosphorylation of c-Jun-terminal-N-kinase and c-Jun. Subsequently, the effect of Aza was investigated in aneurysm formation in the angiotensin II aneurysm mouse model in apolipoprotein E-deficient mice. We demonstrated that Aza decreases de novo aortic aneurysm formation from an average aneurysm severity score of 2.1 (control group) to 0.6 (Aza group), and that Aza effectively delays aorta pathology in a progression experiment, resulting in a reduced severity score from 2.8 to 1.7 in Aza-treated mice. In line with the in vitro observations, Aza-treated mice showed less c-Jun-terminal-N-kinase activation in ECs and reduced leukocyte influx in the aortic wall. CONCLUSIONS: The immunosuppressive drug Aza has an anti-inflammatory effect and in ECs inhibits Rac1 and c-Jun-terminal-N-kinase activation, which may explain the protective effect of Aza in aneurysm development and, most importantly for clinical implications, aneurysm severity.


Assuntos
Aneurisma Aórtico/prevenção & controle , Azatioprina/farmacologia , Células Endoteliais/efeitos dos fármacos , Imunossupressores/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Neuropeptídeos/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas rac1 de Ligação ao GTP/antagonistas & inibidores , Angiotensina II , Animais , Anti-Inflamatórios/farmacologia , Aneurisma Aórtico/induzido quimicamente , Aneurisma Aórtico/enzimologia , Aneurisma Aórtico/genética , Aneurisma Aórtico/imunologia , Aneurisma Aórtico/patologia , Ruptura Aórtica/enzimologia , Ruptura Aórtica/imunologia , Ruptura Aórtica/prevenção & controle , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Modelos Animais de Doenças , Progressão da Doença , Células Endoteliais/enzimologia , Células Endoteliais/imunologia , Ativação Enzimática , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Mediadores da Inflamação/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mercaptopurina/metabolismo , Camundongos , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Monócitos/imunologia , Neuropeptídeos/metabolismo , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo
18.
Blood Adv ; 7(19): 5868-5876, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37428870

RESUMO

Activated eosinophils are described to release eosinophil extracellular traps (EETs), which consist of the cell's DNA covered with granule-derived antimicrobial peptides. Upon stimulation of eosinophils with the known EET-inducers phorbol 12-myristate 13-acetate, monosodium urate crystals, or Candida albicans, we observed that their plasma membrane became compromised, resulting in accessibility of the nuclear DNA for staining with the impermeable DNA dye Sytox Green. However, we did not observe any DNA decondensation or plasma membrane rupture by eosinophils, which sharply contrasts with neutrophil extracellular trap (NET) formation and the subsequent cell death known as NETosis. Neutrophil elastase (NE) activity is thought to be essential for the cleavage of histones and chromatin decondensation during NETosis. We observed that the neutrophils of a patient with a mutation in ELANE, leading to congenital neutropenia and NE deficiency, were unable to undergo NETosis. Taken together, we may suggest that the natural absence of any NE-like proteolytic activity in human eosinophils explains why EET formation is not observed, even when eosinophils become positive for an impermeable DNA dye in response to stimuli that induce NETosis in neutrophils.


Assuntos
Armadilhas Extracelulares , Humanos , Armadilhas Extracelulares/metabolismo , Neutrófilos/metabolismo , Histonas/metabolismo , DNA/metabolismo , Membrana Celular/metabolismo
19.
Front Immunol ; 14: 1183180, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37261342

RESUMO

Cancer is one of the leading causes of death worldwide. Treatment outcome is largely dictated by the tumor type, disease stage, and treatment success rates, but also by the variation among patients in endogenous anti-tumor responses. Studies indicate that the presence of neutrophils in the tumor microenvironment is associated with a worse patient outcome due to their ability to suppress local anti-tumor T cell activity. Our previous studies investigated the mechanisms by which neutrophils suppress and damage T cells to become smaller in size (small T cells), debilitating their effector activities. Several studies indicate a role for tumor-associated macrophages in scavenging damaged or dead cells. We hypothesized that the observed lack of small T cells in the TME by confocal microscopy is due to immediate uptake by macrophages. In this study, we confirmed that indeed only the smaller, damaged T cells are taken up by macrophages, once serum-opsonized. Damaged T cells opsonized with complement factor C3 fragments were phagocytosed by macrophages, resulting in almost instantaneous and highly efficient uptake of these small T cells. Inhibition of the complement receptors CR1, CR3 and CR4 expressed by macrophages completely blocked phagocytosis. By contrast, actively proliferating T cells (large T cells) were neither impaired in neutrophil-MDSC activity nor opsonized for phagocytosis by macrophages. Rapid removal of damaged T cells suggests a role of complement and macrophages within the tumor microenvironment to clear suppressed T cells in cancer patients.


Assuntos
Macrófagos , Linfócitos T , Humanos , Receptores de Complemento 3b , Receptores de Complemento/fisiologia , Complemento C3
20.
Blood Adv ; 7(23): 7169-7183, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-37792794

RESUMO

E-cadherin is a crucial regulator of epithelial cell-to-cell adhesion and an established tumor suppressor. Aside epithelia, E-cadherin expression marks the erythroid cell lineage during human but not mouse hematopoiesis. However, the role of E-cadherin in human erythropoiesis remains unknown. Because rat erythropoiesis was postulated to reflect human erythropoiesis more closely than mouse erythropoiesis, we investigated E-cadherin expression in rat erythroid progenitors. E-cadherin expression is conserved within the erythroid lineage between rat and human. In response to anemia, erythroblasts in rat bone marrow (BM) upregulate E-cadherin as well as its binding partner ß-catenin. CRISPR/Cas9-mediated knock out of E-cadherin revealed that E-cadherin expression is required to stabilize ß-catenin in human and rat erythroblasts. Suppression of ß-catenin degradation by glycogen synthase kinase 3ß (GSK3ß) inhibitor CHIR99021 also enhances ß-catenin stability in human erythroblasts but hampers erythroblast differentiation and survival. In contrast, direct activation of ß-catenin signaling, using an inducible, stable ß-catenin variant, does not perturb maturation or survival of human erythroblasts but rather enhances their differentiation. Although human erythroblasts do not respond to Wnt ligands and direct GSK3ß inhibition even reduces their survival, we postulate that ß-catenin stability and signaling is mostly controlled by E-cadherin in human and rat erythroblasts. In response to anemia, E-cadherin-driven upregulation and subsequent activation of ß-catenin signaling may stimulate erythroblast differentiation to support stress erythropoiesis in the BM. Overall, we uncover E-cadherin/ß-catenin expression to mark stress erythropoiesis in rat BM. This may provide further understanding of the underlying molecular regulation of stress erythropoiesis in the BM, which is currently poorly understood.


Assuntos
Anemia , beta Catenina , Humanos , Ratos , Camundongos , Animais , beta Catenina/metabolismo , Eritropoese/fisiologia , Glicogênio Sintase Quinase 3 beta , Caderinas/genética , Caderinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA